Prognostic Factors in Hodgkin Lymphoma

Annarosa Cuccaro, Francesca Bartolomei, Elisa Cupelli, Eugenio Galli, Manuela Giachelia and Stefan Hohaus

Institute of Hematology, Catholic University S. Cuore, Rome, Italy.

Corresponding author: Dr Stefan Hohaus. Istituto di Ematologia, Universita’ Cattolica S. Cuore. L.go A. Gemelli, 1, 00168 ROMA. Tel: -39-06-30154180, Fax: -39-06-35503777. E-mail: stefan.hohaus@rm.unicatt.it

Published: July 5, 2014
Received: June 3. 2014
Accepted: July 2, 2014
Meditterr J Hematol Infect Dis 2014, 6(1): e2014053, DOI 10.4084/MJHID.2014.053
This article is available on PDF format at:

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Hodgkin lymphoma (HL) is among the neoplastic diseases that has the best long-term outcome after cytotoxic treatment. Cure rates approach 80-90%; however, 15-20% of patients will be resistant to therapy (primary refractory) or relapse after treatment. Prognostic factors should help to stratify treatment according to the risk profile and identify patients at risk for failure. Significance of prognostic factors partly depends on the efficacy of the treatments administered, since new effective therapies can variably counterbalance the adverse effects of some unfavorable clinical determinants. As a consequence, some prognostic factors thought to be important in the past may become meaningless when modern successful therapies are used. Therefore, the value of prognostic factors has to be updated periodically, and then adapted to new emerging biomarkers. Besides the prognostic role of PET imaging, tissue and circulating biomarkers, as the number of tumor-infiltrating macrophages, cytokine and chemokine levels and profiling of circulating nucleic acids (DNA and microRNAs) have shown promise.

Introduction

Treatment of Hodgkin lymphoma (HL) is an indubitable one of the greatest success stories of medical oncology in the 20th century. Cure rates approach 80-90% of patients, and HL is among the neoplastic diseases that have the best long-term outcome after cytotoxic treatment. However, 15-20% of patients will be resistant to therapy (primary refractory) or relapse after treatment, usually in the first two years. This review will analyze the prognostic factors that can identify patients at risk. Since outcome of patients is determined not only by disease characteristics but also by the risk of short- and long-term sequelae of the treatment, which can even outnumber the events of disease recurrence, the identification of risk factors for secondary events will be increasingly important to tailor the therapy and thus avoiding potential harmful treatments in individuals at risk.
In a simplified way, prognostic factors can be divided into areas that are related to the disease, factors related to the patient as a host for the disease and to the therapy (Figure 1). Interactions between these areas exist. The genetic background of the patient is a host factor that modulates the metabolism of cytotoxic drugs, and as a consequence alters the response and side effects of the treatment.


Figure 1 Figure 1. Prognostic factors can be divided into areas that are related to the disease, factors related to the patient as a host for the disease and to the therapy. Interactions between these areas exist. The genetic background of the patient is a host factor that modulates the metabolism of cytotoxic drugs, and as a consequence alters the response and side effects of the treatment.

In this review, we will only briefly discuss the prognostic relevance of pathological and immunological features of HL, and not consider PET imaging, that has evolved into the most exciting tool to evaluate the prognosis in HL in recent years. This topic will be covered with another review in this issue of the journal. Many prognostic factors, used in standard clinical practice, have been known for a long time. These factors often reflect disease burden and disease activity that is related to the inflammatory microenvironment. Biomarkers described in recent years are indicators of the disease activity as well, but they describe this activity in a more sophisticated, accurate and pathogenetically more relevant way. Often these new prognostic factors still need validation, but they may eventually substitute for classical clinical factors.[1]

Tumor Burden: Stage and Bulk

Extension of disease and tumor burden is indubitable the most important disease characteristic, that is used to stratify treatment strategies (Figure 2). Staging according to the Ann Arbor system is part of clinical routine for more than 40 years.[2] In limited stage disease, the presence of bulky disease detected on chest radiography or CT at staging is considered a negative predictor of outcome. The presence of a bulky tumor is one of the risk factors in the European Organization for Research and Treatment of Cancer/Groupe d’étude des Lymphomes de l’adulte (EORTC/GELA) and the German Hodgkin Study Group (GSHG) stratification scores for HL.[3] By contrast, in advanced stage disease, the presence of a bulky tumor is not a risk factor in the international prognostic score (IPS) for HL.[4] Since measurement of bulk is limited to the single largest mass, it could underestimate the total tumor burden in patients with diffuse disease. Newer methods to measure tumor burden with CT volume or metabolic tumor volume may give more precise estimation of the tumor volume.[5-7] Normalization of the tumor mass to the body surface (the relative tumor burden) yields a parameter with a reliable prediction for tumor control modulated by the use of chemotherapy regimens with different intensity.[7] The complexity of the evaluation of all lesions in any scan slice with subtraction of normal structures that are present in the tumor tissue, and approximation for bone marrow involvement has limited a wider application of this type of evaluation. Therefore, an indirect estimate was derived from a few staging parameters and demonstrated sufficient statistical reliability when compared with the direct measure of rTB.[8] The equation {Estimated rTB = −4.3 + 8.3 × IPI2+ 22.7 × [number of involved sites (+3 if bulky mass is present)]} was recently proposed for investigational and clinical uses when the direct measurement cannot be performed.


Figure 2 Figure 2. Risk factors in the European Organization for Research and Treatment of Cancer/Groupe d’étude des Lymphomes de l’adulte (EORTC/GELA) and the German Hodgkin Study Group (GSHG) stratification scores for HL.

Spread of HL beyond its lymph node microenvironment to extralymphatic organs is associated with inferior outcome. In limited stage disease involvement of an extranodal site is defined as a risk factor by the GSHG scoring system. In patients with advanced-stage disease, diffuse organ involvement defining stage IV disease is an independent risk factor in the IPS.[9]

Age

Age is the most important factor when overall survival is analyzed, and remains an independent factor also for progression-free survival. It impacts on prognosis in at least two ways: On one hand, it is intrinsically associated with HL biology and, on the other hand, older age often is associated with co-morbidity and reduced tolerability of chemotherapeutic regimens used in younger patients. HL epidemiology is characterized by a bimodal age distribution. Following the peak in young adults in their twenties, there is a second increase in the incidence, in particular in males, after the age of 50-55 years. When compared to other hematological neoplastic diseases, that usually set the cut-point to define elderly patients at 60 years, the prognostic cut-point in HL is shifted versus a younger age.
In the International Prognostic Score for patients with advanced stage disease the cut-point is age of 45 years, the EORTC lists age more than 50 years as a risk factor for patients with limited stage disease. Older age associates with a higher frequency of the mixed cellularity histotype and presence of EBV in the neoplastic cells, when compared to younger patients.[10] EBV-association appears to be a prognostic factor that is limited to the elderly patients.[11-13] It is hypothesized that loss of immunological control of EBV-infected cells might contribute to the development of EBV-associated HL in the elderly. Aging of the immune system (immunosenescence) is characterized by reduced function of the adaptive immune response that includes T and B cell function. Studies are required to address the question whether immunosenescence is a mechanism in the pathogenesis of elderly HL, and whether this will contribute to the negative effect of age on prognosis.
Therapy of HL in the elderly is often complicated by toxic side effects of chemotherapy. Standard treatment with ABVD is often not recommended for patients older than 70 years. Bleomycin leads to frequent incidence of pulmonary toxicity in the elderly. In a recent report, the incidence of bleomycin lung toxicity was 32% with a 25% mortality.[14] Intensified regimens as the BEACOPP-dose escalated regimen are not recommended for patients with advanced-stage HL over 60 years.[15] However, even in patients over 50 years with reduced performance status, mortality of BEACOPP-dose escalated increases to 13.3%.[16] Therapy of elderly patients with HL remains a challenge, and effective regimen with acceptable toxicity profiles is still lacking. The availability of antibody-drug conjugates, as Brentuximab may be major step forward.

Gender

Males with HL have a poorer outcome than females. This effect of gender is not limited to HL. As well, female patients with follicular lymphoma and diffuse large B cell lymphoma fare better than their male counterparts. On the mechanism of the gender effect on prognosis in HL, one can only speculate, but it could influence prognosis in at least two ways. A preponderance of male gender is observed in elderly patients, and as a consequence males have more often unfavorable disease characteristics. Another mechanism for the gender effect in lymphoma may be due to differences in pharmacokinetics. Female patient with HL experiences more hematological toxicity, especially more severe leucopenia, probably due to gender difference in metabolism of cytotoxic drugs of the ABVD regimen.[17] Moreover, hematological toxicity has been associated with a more favorable outcome.

B-Symptoms

Constitutional symptoms defined by unexplained fever >38°C, drenching night sweats and weight loss >10% of the weight are a presenting sign in about 10-25% of patients with limited stage disease, and up to 70% of patients with advanced stage disease.[9] Among the symptoms, isolated night sweats do not appear to be associated with inferior outcome. The presence of B-symptoms is a risk factor, in particular in stage II bulky disease, that is not considered to be a limited stage disease by the German Hodgkin study group when B-symptoms are present.
B-symptoms are due to the production of pro-inflammatory cytokines by the Hodgkin tumor tissue, in particular IL-1, TNF-alpha, and IL-6.[1] B-symptoms are associated with a variety of other laboratory abnormalities and patients’ characteristics, and in multivariate analyses it has therefore often been removed in final models, as in the IPS.

Anemia

Anemia is a frequent finding at HL diagnosis and is present in about 40% of patients. It is usually a mild to moderate normocytic anemia, with the characteristics of anemia of inflammation. Cut-off point for prognosis in the IPS is a hemoglobin level of 10.5 g/dl, and this is independent of gender.
We demonstrated that elevated IL-6 levels correlate with hemoglobin levels and that IL-6 levels correlate with levels of hepcidin, an acute phase reactant and a major regulator of iron metabolism.[18] Therefore, anemia is linked to the inflammatory activity of the HL microenvironment, and this might explain its big prognostic impact.
Anemia of inflammation is characterized by alterations in iron metabolism. Elevated production of hepcidin blocks the release of iron from the intestine and iron stores in the reticuloendothelial system that results in increasing levels of ferritin. Elevated ferritin levels have been described in HL and have been associated to prognosis four decades ago.[19] The accumulation of iron also in the HL microenvironment can have biologic effects on cell function and induce cell damage by induction of reactive oxygen species (ROS) that interfere with the function of macromolecules as DNA, and proteins.

The White Blood Cells: Leukocytosis, Lymphopenia, Monocytosis

Alterations of the counts and composition of the white blood cells in peripheral blood are often at diagnosis in HL and well known prognostic factors. Typical alterations in WBC counts include leukocytosis with neutrophilia, lymphocytopenia, either relative or absolute, and monocytosis.
In the IPS the prognostic cut point for white blood cell count is set at 15000/microL, for lymphocytopenia it is 600/microL or less than 8%.[9] More recently, the monocyte count, in particular in relation to the lymphocyte count has been reported to be a prognostic factor in HL and other lymphomas.[20] In a case cohort of 474 patients with HL observed from 1974 to 2010, monocyte count of >900/microL was associated with inferior progression-free and overall survival. The impact of the monocyte counts on prognosis became particularly evident when the ratio between lymphocytes and monocytes (ALM ratio) was < 1.1. As the number of macrophages in the HL tissue is strongly associated with prognosis, the question arises whether the number of monocytes in PB and the number of tumor-associated macrophages (TAM) are correlated.

Albumin

Low levels of serum albumin are associated with a worse prognosis in many hematological neoplasias, including HL. The IPS score defines albumin levels of 4.0 g/dl as cut-point. Albumin is produced by the liver, and about 12-20% of the protein synthesis capacity of the liver is dedicated to albumin production. Albumin synthesis is reduced when synthesis of acute-phase proteins is stimulated by IL-6 or when availability of amino acids is decreased due to reduced nutritional status. Albumin levels inversely correlate to IL-6, TNF alpha and IL1-RA.[1]

The Erythrocyte Sedimentation Rate

The erythrocyte sedimentation rate, albeit its nonspecific character is one of the oldest prognostic factors for HL. It is still in use to define early stage HL as favorable or unfavorable. The EORTC and GSHG set the cut-point to 30 mm/h for patients with B –Symptoms and 50 mm/h for patients without B-symptoms. The ESR is increased in many diseases, in particular in those with an inflammatory reaction. The ESR can be altered by many variables, as the erythrocyte count and the protein composition in the plasma, in particular increased levels of fibrinogen, acute phase proteins and gamma globulins can increase the ESR. As these parameters are as well prognostic markers in HL, the ESR does often not maintain its value in multivariate analysis.

Beta2-microglobulin

B2M is a component of the HLA-I antigen and present on the surface of nearly all nucleated cells in the body. In healthy people, it is produced at a constant rate and eliminated in the kidney where free glomerular filtration is followed by tubular re-adsorption. Lymphocytes are the main production site of b2M, and inflammatory cytokines stimulate the production of b2M, and increased levels of b2M can be due to increased release from immune system activation or proliferation or decreased renal clearance. It is a prognostic marker in many lymphomas, including HL. Elevated levels of B2M can be found in 5-30% of patients at diagnosis, depend on the stage, and have been found to be associated with the relapse.[21-23]

Biohumoral Factors: IL-10, IL-6, sCD30, TNF, TARC

A large array of cytokines can be detected at increased levels in peripheral blood in HL. These are produced both by the HRS cells and the surrounding microenvironment. The prognostic significance of cytokine levels has been studied for more than 20 years in HL, and the most frequently studied cytokines are IL-10, IL-6, TNF alpha and its soluble receptors, and more recently the chemokine TARC. IL-10 is of particular interest in the immunopathogenesis of HL, as it is supposed to play an important role in the shift of T cell function from Th1 to Th2 and Treg functional state. IL-10 levels are elevated in about 40-50% of patients, and associate with inferior outcome.1,[24-30] IL-10 levels appear to be higher in EBV-associated HL.[29]
IL-6 is a pro-inflammatory cytokine that is associated with some clinical and laboratory manifestations of HL, as, B-symptoms, anemia, and low albumin levels.[31] IL-6 can induce the production of hepcidin, a major regulator of iron metabolism and mediator for anemia of inflammation or chronic disease, characterized by iron-restriction. We showed that IL-6 – hepcidin axis is active in anemia associated with HL, but that other IL-6-induced, hepcidin-independent mechanisms probably play a role.[30]
The circulating CD30 antigen sCD30, is thought to be shed form the CD30+ HRS cells, and represents, therefore, at least theoretically, an ideal tumor marker for the neoplastic cells. sCD30 levels are increased in about 25-30 % of patients with HL, and, levels above 100 -200 U/ml associate with worse outcome.[32]
Ma et al.[33] used a proteomic approach to screen for proteins in plasma at HL diagnosis to identify new protein biomarkers. The most promising biomarkers appeared to be TARC (thymus and activation-regulated chemokine), a chemokine that is important for attracting immune cells with specific functions to the microenvironment.
The chemokine TARC has recently attracted more interest as it plays a central role in the composition of the microenvironment attracting Th2 and Treg cells. TARC levels are elevated in the vast majority of patients with HL at diagnosis, and rapidly turn to normal during treatment.[34-35] Preliminary data indicate an association between changes in TARC concentration in plasma and therapy outcome. Whether this early change can be a marker to evaluate response has to be addressed in larger studies.
Casanovas and colleagues developed a prognostic score based on different cytokine levels.[1] The score included IL-6, sCD30 and TNFR1 and was more predictive than standard clinical score. While this work is of high interest, these data need confirmation on independent data sets and in relation to the results of early or interim PET.

Prognostic Relevance of Characteristics of HRS Cells

The number and atypia of HRS cells together with the degree of cellularity in the nodules and the amount of sclerosis are the characteristics for the separation of nodular sclerosis (NS) into grade 1 and grade 2 according to the British National Lymphoma Investigation (BNLI)[].[36] NS grade 2 typically is more aggressive, and has an inferior outcome. However, difficulties to reproduce this classification has resulted in conflicting data and limited the widespread use of this classification.[37-38]
Several studies indicated that BCL-2 expression in HRS cells is associated with an inferior prognosis.[39-41] However, the relationship between BCL-2 expression and patient outcome in HL remains controversial because other studies have not demonstrated the same correlation between bcl-2 expression and failure-free survival.[42] Similarly, the association of p53 with patient outcomes in HL remains controversial[40-42] although more studies suggest a prognostic role for BCL-2 than for p53.

Prognostic Relevance of the Tumor Microenvironment

HL is characterized by an expansion of T cells with a T helper2 and T regulatory phenotype in the microenvironment. However, both immunohistochemistry and gene expression studies indicate that high numbers of T cells with a cytotoxic phenotype and low numbers of FOXP-3 + T reg cells in the microenvironment are associated with inferior outcome.[43-45] A number of other components in the microenvironment as, B cells and eosinophils have been reported to be associated with prognosis.[46-47] However, this information is not part of the routine evaluation for the prognostic purpose.
A more recent tissue biomarker is the number of tumor-infiltrating macrophages, identified by immunohistochemical staining for the CD68 antigen,[48] which is a relatively simple tissue biomarker of gaining widespread interest.[49] However, not all studies could confirm the prognostic impact of the count of tumor-associated macrophages in HL. Further studies are needed to determine the optimal antigen (e.g. CD68 versus CD163), anti-CD68 antibody clone (e.g. KP1 versus PGM1) and scoring thresholds (e.g. manual versus computer-assisted) for detecting HL associated macrophages.49 The Vancouver group developed a 23-gene outcome predictor that was superior to the IPS and to CD68 immunohistochemistry.

Circulating DNA of Cellular and Viral (EBV) Origin

Cell-free DNA of cellular and viral origin can be detected in the plasma of patients with HL at diagnosis.[50-51] Cell-free DNA is released from the tumor tissue, and levels correlate to disease activity.[52] Cell-free DNA is probably released both by the tumor cells and the surrounding microenvironment. The identification of recurrent mutations in patients with HL opens the possibility to develop sensitive techniques to detect these mutations in the cell-free DNA fraction as specific tumor markers in the peripheral blood.[52-53] Patients with cell-free DNA levels above the normal range have an inferior event-free survival.[50] In the same line, EBV-DNA can be detected in the plasma of patients with HL, and represents a marker for the activity of EBV-associated HL.[54] It is important to underline that detection of EBV in plasma, but not in the mononuclear cell fraction is associated with the EBV-status in HL.[51,54] We and others have shown that the presence and level of EBV-DNA is a prognostic marker. [51,54-55]

Genetic Background

Genome-wide association studies (GWAS) on large cohorts of patients with HL have defined the role of polymorphic germ line variants as a risk factor for the development of HL.[56-58] These studies commonly identified a locus on chromosome 6 in the HLA region as a highly significant risk allele for HL. Other single nucleotide polymorphisms, in other HLA regions and cytokine genes, as IL-13 have also been associated with HL risk. About the role of the genetic background as a factor that can modulate the response to treatment and outcome of patients with HL, no results of GWAS are available. Using a target gene approach, we and others reported on the prognostic impact of SNPs in HL. Most of genes studied were involved in the metabolism of cytotoxic drugs, as detoxification enzymes, and immunoregulatory genes, given the pivotal role of immunological alterations and interactions in the pathology of the disease.[59-66] In particular, we found deletions of GSTT1 and GSTM1 and a variant in the GSTP1 gene (Val105Ile), which reduces the enzymatic activity, be associated to a better outcome.[59-60] These data could only be partially confirmed on an independent cohort of patients with advanced HL included in multicenter trial.[67] Other studies reported on the prognostic impact of genes coding for enzymes involved in drug metabolism are and UGT1A1 and GSTA1.[61,64] We reported that carriers of variants in the promoter region of the IL-6 and IL-10 genes that are supposed to influence gene expression were associated with prognosis in HL.[68] Validation on independent and large patient cohorts is needed before the germ line variants in the genetic background can be clinically used to modulate the treatment.

Prognostic Scores

Clinical and laboratory parameters have been combined into different prognostic scoring systems. Patients with limited stage disease are traditionally divided into a favorable/unfavorable group according to the presence of risk factors defined with some minor differences by the GSHG and EORTC (Figure 2).[69] Both GSHG and EORTC use this classification to design treatment protocols that adapt therapy intensity according to the risk group.
Hasenclever et al. used the data on 5141 patients with advanced stage disease treated with ABVD or COPP-ABVD to develop a 7-point score (international prognostic score, IPS). In order to stratify patients with advanced stage disease, patients are divided into two risk groups (IPS 0-2 vs. IPS 3-5). However, stratification of therapy according to Hasenclever score has not entered clinical routine.
Risk scores have also been developed for patients with relapsed disease. Risk factors as early relapse within 12 months, presence of B-symptoms and extranodal disease are the most important clinical factors, as anemia appears to be the most significant laboratory anomaly to predict poor outcome in relapsed patients.[67,69]

Conclusions

In conclusion, a plethora of prognostic factors is available in HL. Traditional clinical and laboratory prognostic factors often represent a surrogate marker for biological characteristics that often are not included in the standard evaluation. There is no current consensus on how to integrate these biological markers with accepted clinical prognostic risk factors into prognostic scores or how to use this information to adapt treatment. It remains a challenge to identify the best parameters to predict prognosis in the single patient and identify the still significant group of patients for whom standard treatment is not sufficient.

References

  1. Casasnovas RO, Mounier N, Brice P, Divine M, Morschhauser F, Gabarre J, Blay JY, Voillat L, Lederlin P, Stamatoullas A, Bienvenu J, Guiguet M, Intrator L, Grandjean M, Brière J, Ferme C, Salles G; Groupe d'Etude des Lymphomes de l'Adulte. Plasma cytokine and soluble receptor signature predicts outcome of patients with classical Hodgkin's lymphoma: a study from the Groupe d'Etude des Lymphomes de l'Adulte. J Clin Oncol. 2007; 25:1732-40. http://dx.doi.org/10.1200/JCO.2006.08.1331 PMid:17389336      
  2. Carbone PP, Kaplan HS, Musshoff K, Smithers DW and Tubiana M. Report of the Committee on Hodgkin's Disease Staging Classification. Cancer Research. 1971; 31:1860- 1861. PMid:5121694      
  3. Eichenauer DA, Engert A, Dreyling M; ESMO Guidelines Working Group. Hodgkin's lymphoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2011; 22 Suppl 6:55-8. http://dx.doi.org/10.1093/annonc/mdr378 PMid:21908505      
  4. Hasenclever D, Diehl V. A prognostic score for advanced Hodgkin’s disease. International prognostic factors project on advanced Hodgkin’s disease. N Engl J Med. 1998; 339:1506-14. http://dx.doi.org/10.1056/NEJM199811193392104 PMid:9819449       
  5. Gobbi PG, Bassi E, Bergonzi M, Merli F, Coriani C, Iannitto E, Luminari S, Polimeno G, Federico M. Tumour burden predicts treatment resistance in patients with early unfavourable or advanced stage Hodgkin lymphoma treated with ABVD and radiotherapy. Hematol Oncol. 2012; 30:194-9. http://dx.doi.org/10.1002/hon.1024 PMid:22271092       
  6. Berkowitz A, Basu S, Srinivas S, Sankaran S, Schuster S, Alavi A. Determination of whole-body metabolic burden as a quantitative measure of disease activity in lymphoma: a novel approach with fluorodeoxyglucose-PET. Nucl Med Commun. 2008; 29:521-6. http://dx.doi.org/10.1097/MNM.0b013e3282f813a4 PMid:18458598       
  7. Gobbi PG. Tumor burden in Hodgkin’s lymphoma: Much more than the best prognostic factor. Critical Reviews in Oncology/Hematology 2014; 90:17-23. http://dx.doi.org/10.1016/j.critrevonc.2013.11.002 PMid:24290380       
  8. Gobbi PG, Bergonzi M, Bassi E, Merli F, Coriani C, Stelitano C, Iannitto E, Federico M. Tumor burden in Hodgkin’slymphoma can be reliably estimated from a few staging parameters. Oncol Rep 2012; 28:815-20. PMid:22752083       
  9. Hasenclever D, Diehl V. A prognostic score for advanced Hodgkin's disease. International Prognostic Factors Project on Advanced Hodgkin's Disease. N Engl J Med. 1998; 339:1506-14. http://dx.doi.org/10.1056/NEJM199811193392104 PMid:9819449       
  10. Massini G, Siemer D, Hohaus S. EBV in Hodgkin lymphoma. Mediterr J Hematol Infect Dis. 2009; 1:e2009013. PMid:21416003 PMCid:PMC3033177  
  11. Jarrett RF, Stark GL, White J, Angus B, Alexander FE, Krajewski AS, Freeland J, Taylor GM, Taylor PR; Scotland and Newcastle Epidemiology of Hodgkin Disease Study Group. Impact of tumor Epstein-Barr virus status on presenting features and outcome in age-defined subgroups of patients with classic Hodgkin lymphoma: a population-based study. Blood. 2005;106:2444-51. http://dx.doi.org/10.1182/blood-2004-09-3759 PMid:15941916       
  12. Keegan TH, Glaser SL, Clarke CA, Gulley ML, Craig FE, Digiuseppe JA, Dorfman RF, Mann RB, Ambinder RF. Epstein-Barr virus as a marker of survival after Hodgkin's lymphoma: a population-based study. J Clin Oncol. 2005; 23:7604-13. http://dx.doi.org/10.1200/JCO.2005.02.6310 PMid:16186595       
  13. Diepstra A, van Imhoff GW, Schaapveld M, Karim-Kos H, van den Berg A, Vellenga E, Poppema S. Latent Epstein-Barr virus infection of tumor cells in classical Hodgkin's lymphoma predicts adverse outcome in older adult patients. J Clin Oncol. 2009; 27:3815-21. http://dx.doi.org/10.1200/JCO.2008.20.5138 PMid:19470931       
  14. Evens AM, Helenowski I, Ramsdale E, Nabhan C, Karmali R, Hanson B, Parsons B, Smith S, Larsen A, McKoy JM, Jovanovic B, Gregory S, Gordon LI, Smith SM. A retrospective multicenter analysis of elderly Hodgkin lymphoma: outcomes and prognostic factors in the modern era. Blood. 2012;119:692-5. http://dx.doi.org/10.1182/blood-2011-09-378414 PMid:22117038       
  15. Diehl V, Stein H, Hummel M, Zollinger R, Connors JM. Hodgkin's lymphoma: biology and treatment strategies for primary, refractory, and relapsed disease. Hematology Am Soc Hematol Educ Program. 2003:225-47. http://dx.doi.org/10.1182/asheducation-2003.1.225    
  16. Wongso D, Fuchs M, Plutschow A, Klimm B, Sasse S, Hertenstein B, Maschmeyer B, Vieler T, Duhrsen U, Lindemann W, Aulitzky W, Diehl V, Borchmann P, and Engert A. Treatment-Related Mortality in Patients With Advanced-Stage Hodgkin Lymphoma: An Analysis of the German Hodgkin Study Group. J Clin Oncol. 2013; 31:2819-24. http://dx.doi.org/10.1200/JCO.2012.47.9774 PMid:23796987       
  17. Klimm B, Reineke T, Haverkamp H, Behringer K, Eich HT, Josting A, Pfistner B, Diehl V, Engert A; German Hodgkin Study Group. Role of hematotoxicity and sex in patients with Hodgkin's lymphoma: an analysis from the German Hodgkin Study Group. J Clin Oncol. 2005; 23:8003-11. http://dx.doi.org/10.1200/JCO.2005.205.60 PMid:16204002       
  18. Hohaus S, Massini G, Giachelia M, Vannata B, Bozzoli V, Cuccaro A, D'Alo' F, Larocca LM, Raymakers RA, Swinkels DW, Voso MT, Leone G. Anemia in Hodgkin's lymphoma: the role of interleukin-6 and hepcidin. J Clin Oncol. 2010; 28:2538-43. http://dx.doi.org/10.1200/JCO.2009.27.6873 PMid:20406921       
  19. Hohaus S, Giachelia M, Cuccaro A, Voso MT, Leone G. Iron in Hodgkin's lymphoma. Crit Rev Oncog. 2013; 18:463-9. http://dx.doi.org/10.1615/CritRevOncog.2013007765    
  20. Porrata LF, Ristow K, Colgan JP, Habermann TM, Witzig TE, Inwards DJ, Ansell SM, Micallef IN, Johnston PB, Nowakowski GS, Thompson C, Markovic SN. Peripheral blood lymphocyte/monocyte ratio at diagnosis and survival in classical Hodgkin's lymphoma. Haematologica. 2012; 97:262-9. http://dx.doi.org/10.3324/haematol.2011.050138 PMid:21993683 PMCid:PMC3269488  
  21. Dimopoulos MA, Cabanillas F, Lee JJ, Swan F, Fuller L, Allen PK, Hagemeister FB. Prognostic role of serum beta 2-microglobulin in Hodgkin's disease. J Clin Oncol. 1993; 11:1108-11. PMid:8501496       
  22. Chronowski GM, Wilder RB, Tucker SL, Ha CS, Sarris AH, Hagemeister FB, Barista I, Hess MA, Cabanillas F, Cox JD. An elevated serum beta-2-microglobulin level is an adverse prognostic factor for overall survival in patients with early-stage Hodgkin disease. Cancer. 2002; 95:2534-8. http://dx.doi.org/10.1002/cncr.10998 PMid:12467067       
  23. Vassilakopoulos TP, Nadali G, Angelopoulou MK, Siakantaris MP, Dimopoulou MN, Kontopidou FN, Karkantaris C, Kokoris SI, Kyrtsonis MC, Tsaftaridis P, Pizzolo G, Pangalis GA. The prognostic significance of beta(2)-microglobulin in patients with Hodgkin's lymphoma. Haematologica. 2002; 87:701-8. PMid:12091120       
  24. Sarris AH1, Kliche KO, Pethambaram P, Preti A, Tucker S, Jackow C, Messina O, Pugh W, Hagemeister FB, McLaughlin P, Rodriguez MA, Romaguera J, Fritsche H, Witzig T, Duvic M, Andreeff M, Cabanillas F. Interleukin-10 levels are often elevated in serum of adults with Hodgkin's disease and are associated with inferior failure-free survival. Ann Oncol 1999; 10:433-40. http://dx.doi.org/10.1023/A:1008301602785    
  25. Bohlen H, Kessler M, Sextro M, Diehl V, Tesch H. Poor clinical outcome of patients with Hodgkin's disease and elevated interleukin-10 serum levels. Clinical significance of interleukin-10 serum levels for Hodgkin's disease. Ann Hematol. 2000; 79:110-113. http://dx.doi.org/10.1007/s002770050564 PMid:10803931      
  26. Axdorph U, Sjöberg J, Grimfors G, Landgren O, Porwit-MacDonald A, Björkholm M. Biological markers may add to prediction of outcome achieved by the International Prognostic Score in Hodgkin's disease. Ann Oncol. 2000; 11:1405-1411. http://dx.doi.org/10.1023/A:1026551727795 PMid:11142480       
  27. Viviani S, Notti P, Bonfante V, Verderio P, Valagussa P, Bonadonna G. Elevated pretreatment serum levels of Il-10 are associated with a poor prognosis in Hodgkin's disease, the milan cancer institute experience. Med Oncol. 2000; 17:59-63. http://dx.doi.org/10.1007/BF02826218 PMid:10713662       
  28. Vassilakopoulos TP, Nadali G, Angelopoulou MK, Siakantaris MP, Dimopoulou MN, Kontopidou FN, Rassidakis GZ, Doussis-Anagnostopoulou IA, Hatzioannou M, Vaiopoulos G, Kittas C, Sarris AH, Pizzolo G, Pangalis GA. Serum interleukin-10 levels are an independent prognostic factor for patients with Hodgkin's lymphoma. Haematologica. 2001;86:274-281. PMid:11255274       
  29. Herling M1, Rassidakis GZ, Medeiros LJ, Vassilakopoulos TP, Kliche KO, Nadali G, Viviani S, Bonfante V, Giardini R, Chilosi M, Kittas C, Gianni AM, Bonadonna G, Pizzolo G, Pangalis GA, Cabanillas F, Sarris AH. Expression of Epstein-Barr virus latent membrane protein-1 in Hodgkin and Reed-Sternberg cells of classical Hodgkin's lymphoma: associations with presenting features, serum interleukin 10 levels, and clinical outcome. Clin Cancer Res. 2003; 9:2114-2120. PMid:12796376      
  30. Hohaus S, Giachelia M, Massini G, Vannata B, Criscuolo M, Martini M, D'Alo' F, Voso MT, Larocca LM, Leone G. Clinical significance of interleukin-10 gene polymorphisms and plasma levels in Hodgkin lymphoma. Leuk Res. 2009;33:1352-6. http://dx.doi.org/10.1016/j.leukres.2009.01.009 PMid:19201467      
  31. Kurzrock R, Redman J, Cabanillas F, Jones D, Rothberg J, Talpaz M. Serum interleukin 6 levels are elevated in lymphoma patients and correlate with survival in advanced Hodgkin's disease and with B symptoms. Cancer Res. 1993;53:2118-22. PMid:8481913       
  32. Nadali G, Tavecchia L, Zanolin E, Bonfante V, Viviani S, Camerini E, Musto P, Di Renzo N, Carotenuto M, Chilosi M, Krampera M, Pizzolo G. Serum level of the soluble form of the CD30 molecule identifies patients with Hodgkin's disease at high risk of unfavorable outcome. Blood. 1998; 91:3011-6. PMid:9531614       
  33. Ma Y, Visser L, Roelofsen H, de Vries M, Diepstra A, van Imhoff G, van der Wal T, Luinge M, Alvarez-Llamas G, Vos H, Poppema S, Vonk R, van den Berg A. Proteomics analysis of Hodgkin lymphoma: identification of new players involved in the cross-talk between HRS cells and infiltrating lymphocytes. Blood. 2008; 111:2339-46. http://dx.doi.org/10.1182/blood-2007-09-112128 PMid:18070985       
  34. Plattel WJ, van den Berg A, Visser L, van der Graaf AM, Pruim J, Vos H, Hepkema B, Diepstra A, van Imhoff GW. Plasma thymus and activation-regulated chemokine as an early response marker in classical Hodgkin's lymphoma. Haematologica. 2012; 97:410-5. http://dx.doi.org/10.3324/haematol.2011.053199 PMid:22058214 PMCid:PMC3291596  
  35. Jones K, Vari F, Keane C, Crooks P, Nourse JP, Seymour LA, Gottlieb D, Ritchie D, Gill D, Gandhi MK. Serum CD163 and TARC as disease response biomarkers in classical Hodgkin lymphoma. Clin Cancer Res. 2013;19:731-42. http://dx.doi.org/10.1158/1078-0432.CCR-12-2693 PMid:23224400       
  36. Bennett MH, MacLennan KA, Easterling MJ, Vaughan Hudson B, Jelliffe AM, Vaughan Hudson G. The prognostic significance of cellular subtypes in nodular sclerosing Hodgkin's disease: an analysis of 271 non-laparotomised cases (BNLI report no. 22). Clin Radiol. 1983;34:497-501. http://dx.doi.org/10.1016/S0009-9260(83)80148-2    
  37. Van Spronsen DJ, Vrints LW, Hofstra G, Crommelin MA, Coebergh JW, Breed WP. Disappearance of prognostic significance of histopathological grading of nodular sclerosing Hodgkin's disease for unselected patients, 1972-92. Br J Haematol. 1997;96:322-7. http://dx.doi.org/10.1046/j.1365-2141.1997.d01-2010.x PMid:9029020       
  38. Ferry JA, Linggood RM, Convery KM, Efird JT, Eliseo R, Harris NL. Hodgkin disease, nodular sclerosis type. Implications of histologic subclassification. Cancer. 1993;71:457-63. http://dx.doi.org/10.1002/1097-0142(19930115)71:2<457::AID-CNCR2820710229>3.0.CO;2-U    
  39. Rassidakis GZ, Medeiros LJ, Vassilakopoulos TP, Viviani S, Bonfante V, Nadali G, Herling M, Angelopoulou MK, Giardini R, Chilosi M, Kittas C, McDonnell TJ, Bonadonna G, Gianni AM, Pizzolo G, Pangalis GA, Cabanillas F, Sarris AH. BCL-2 expression in Hodgkin and Reed-Sternberg cells of classical Hodgkin disease predicts a poorer prognosis in patients treated with ABVD or equivalent regimens. Blood. 2002; 100:3935-3941. http://dx.doi.org/10.1182/blood.V100.12.3935 PMid:12433696       
  40. Sup SJ, Alema-y CA, Pohlman B, Elson P, Malhi S, Thakkar S, Steinle R, Hsi ED. Expression of bcl-2 in classical Hodgkin's lymphoma: an independent predictor of poor outcome. J Clin Oncol. 2005; 23:3773-3779. http://dx.doi.org/10.1200/JCO.2005.04.358 PMid:15809450        
  41. Smolewski P, Robak T, Krykowski E, Blasi-ska-Morawiec M, Niewiadomska H, Pluzanska A, Chmielowska E, Zambrano O Prognostic factors in Hodgkin's disease: multivariate analysis of 327 patients from a single institution. Clin Cancer Res. 2000; 6:1150-1160. PMid:10741746       
  42. Montalbán C, García JF, Abraira V, González-Camacho L, Morente MM, Bello JL, Conde E, Cruz MA, García-Sanz R, García-Lara-a J, Grande C, Llanos M, Martínez R, Flores E, Méndez M, Ponderós C, Rayón C, Sánchez-Godoy P, Zamora J, Piris MA; Spanish Hodgkin's Lymphoma Study Group. Influence of biologic markers on the outcome of Hodgkin's lymphoma: a study by the Spanish Hodgkin's Lymphoma Study Group. J Clin Oncol. 2004; 22:1664-1673. http://dx.doi.org/10.1200/JCO.2004.06.105 PMid:15117989       
  43. Alvaro T, Lejeune M, Salvadó MT, Bosch R, García JF, Jaén J, Banham AH, Roncador G, Montalbán C, Piris MA. Outcome in Hodgkin's lymphoma can be predicted from the presence of accompanying cytotoxic and regulatory T cells. Clin Cancer Res. 2005; 11:1467-1473. http://dx.doi.org/10.1158/1078-0432.CCR-04-1869 PMid:15746048       
  44. Kelley TW, Pohlman B, Elson P, and Hsi ED. The ratio of FOXP3+ regulatory T cells to granzyme B+ cytotoxic T/NK cells predicts prognosis in classical Hodgkin lymphoma and is independent of bcl-2 and MAL expression. Am J Clin Pathol. 2007; 128:958-65. http://dx.doi.org/10.1309/NB3947K383DJ0LQ2 PMid:18024321       
  45. Sánchez-Aguilera A, Montalbán C, de la Cueva P, Sánchez-Verde L, Morente MM, García-Cosío M, García-Lara-a J, Bellas C, Provencio M, Romagosa V, de Sevilla AF, Menárguez J, Sabín P, Mestre MJ, Méndez M, Fresno MF, Nicolás C, Piris MA, García JF; Spanish Hodgkin Lymphoma Study Group. Tumor microenvironment and mitotic checkpoint are key factors in the outcome of classic Hodgkin lymphoma. Blood. 2006;108:662-8. http://dx.doi.org/10.1182/blood-2005-12-5125 PMid:16551964      
  46. Von Wasielewski R, Seth S, Franklin J, Fischer R, Hübner K, Hansmann ML, Diehl V, Georgii A. Tissue eosinophilia correlates strongly with poor prognosis in nodular sclerosing Hodgkin's disease, allowing for known prognostic factors. Blood. 2000;95:1207-13. PMid:10666192       
  47. Chetaille B, Bertucci F, Finetti P, Esterni B, Stamatoullas A, Picquenot JM, Copin MC, Morschhauser F, Casasnovas O, Petrella T, Molina T, Vekhoff A, Feugier P, Bouabdallah R, Birnbaum D, Olive D, Xerri L. Molecular profiling of classical Hodgkin lymphoma tissues uncovers variations in the tumor microenvironment and correlations with EBV infection and outcome. Blood. 2009; 113:2765-75. http://dx.doi.org/10.1182/blood-2008-07-168096 PMid:19096012       
  48. Steidl C, Lee T, Shah SP, Farinha P, Han G, Nayar T, Delaney A, Jones SJ, Iqbal J, Weisenburger DD, Bast MA, Rosenwald A, Muller-Hermelink HK, Rimsza LM, Campo E, Delabie J, Braziel RM, Cook JR, Tubbs RR, Jaffe ES, Lenz G, Connors JM, Staudt LM, Chan WC, Gascoyne RD. Tumor-associated macrophages and survival in classic Hodgkin's lymphoma. N Engl J Med. 2010; 362:875-85. http://dx.doi.org/10.1056/NEJMoa0905680 PMid:20220182 PMCid:PMC2897174
  49. Steidl C, Farinha P, Gascoyne RD. Macrophages predict treatment outcome in Hodgkin's lymphoma. Haematologica. 2011;96:186-9. http://dx.doi.org/10.3324/haematol.2010.033316 PMid:21282720 PMCid:PMC3031684  
  50. Hohaus S, Giachelia M, Massini G, Mansueto G, Vannata B, Bozzoli V, Criscuolo M, D'Alò F, Martini M, Larocca LM, Voso MT, Leone G. Cell-free circulating DNA in Hodgkin's and non-Hodgkin's lymphomas. Ann Oncol. 2009;20:1408-13. http://dx.doi.org/10.1093/annonc/mdp006 PMid:19465421       
  51. Hohaus S, Santangelo R, Giachelia M, Vannata B, Massini G, Cuccaro A, Martini M, Cesarini V, Cenci T, D'Alo F, Voso MT, Fadda G, Leone G, Larocca LM. The viral load of Epstein-Barr virus (EBV) DNA in peripheral blood predicts for biological and clinical characteristics in Hodgkin lymphoma. Clin Cancer Res. 2011; 17:2885-92. http://dx.doi.org/10.1158/1078-0432.CCR-10-3327 PMid:21478335       
  52. Reichel JB, Eng K, Elemento O, Cesarman E, Roshal M. Exome Sequencing Of Purified Hodgkin Reed-Sternberg Cells Reveals Recurrent Somatic Mutations In Genes Responsible For Antigen Presentation, Chromosome Integrity, Transcriptional Regulation and Protein Ubiquitination. Blood 2013 (ASH Annual Meeting Abstracts), 122, 625.  
  53. Gunawardana J, Chan FC, Telenius A, Woolcock B, Kridel R, Tan KL, Ben-Neriah S, Mottok A, Lim RS, Boyle M, Rogic S, Rimsza LM, Guiter C, Leroy K, Gaulard P, Haioun C, Marra MA, Savage KJ, Connors JM, Shah SP, Gascoyne RD, Steidl C. Recurrent somatic mutations of PTPN1 in primary mediastinal B cell lymphoma and Hodgkin lymphoma. Nat Genet. 2014; 46:329-35. http://dx.doi.org/10.1038/ng.2900 PMid:24531327       
  54. Gandhi MK, Lambley E, Burrows J, Dua U, Elliott S, Shaw PJ, Prince HM, Wolf M, Clarke K, Underhill C, Mills T, Mollee P, Gill D, Marlton P, Seymour JF, Khanna R. Plasma Epstein-Barr virus (EBV) DNA is a biomarker for EBV-positive Hodgkin's lymphoma. Clin Cancer Res. 2006; 12:460-4. http://dx.doi.org/10.1158/1078-0432.CCR-05-2008 PMid:16428487       
  55. Kanakry JA, Li H, Gellert LL, Lemas MV, Hsieh WS, Hong F, Tan KL, Gascoyne RD, Gordon LI, Fisher RI, Bartlett NL, Stiff P, Cheson BD, Advani R, Miller TP, Kahl BS, Horning SJ, Ambinder RF. Plasma Epstein-Barr virus DNA predicts outcome in advanced Hodgkin lymphoma: correlative analysis from a large North American cooperative group trial. Blood. 2013; 121:3547-53. http://dx.doi.org/10.1182/blood-2012-09-454694 PMid:23386127 PMCid:PMC3643756  
  56. Enciso-Mora V, Broderick P, Ma Y, Jarrett RF, Hjalgrim H, Hemminki K, van den Berg A, Olver B, Lloyd A, Dobbins SE, Lightfoot T, van Leeuwen FE, Försti A, Diepstra A, Broeks A, Vijayakrishnan J, Shield L, Lake A, Montgomery D, Roman E, Engert A, von Strandmann EP, Reiners KS, Nolte IM, Smedby KE, Adami HO, Russell NS, Glimelius B, Hamilton-Dutoit S, de Bruin M, Ryder LP, Molin D, Sorensen KM, Chang ET, Taylor M, Cooke R, Hofstra R, Westers H, van Wezel T, van Eijk R, Ashworth A, Rostgaard K, Melbye M, Swerdlow AJ, Houlston RS. A genome-wide association study of Hodgkin's lymphoma identifies new susceptibility loci at 2p16.1 (REL), 8q24.21 and 10p14 (GATA3). Nat Genet. 2010;42:1126-30. http://dx.doi.org/10.1038/ng.696 PMid:21037568       
  57. Cozen W, Li D, Best T, Van Den Berg DJ, Gourraud PA, Cortessis VK, Skol AD, Mack TM, Glaser SL, Weiss LM, Nathwani BN, Bhatia S, Schumacher FR, Edlund CK, Hwang AE, Slager SL, Fredericksen ZS, Strong LC, Habermann TM, Link BK, Cerhan JR, Robison LL, Conti DV, Onel K. A genome-wide meta-analysis of nodular sclerosing Hodgkin lymphoma identifies risk loci at 6p21.32. Blood. 2012; 119:469-75. http://dx.doi.org/10.1182/blood-2011-03-343921 PMid:22086417 PMCid:PMC3257012  
  58. Urayama KY, Jarrett RF, Hjalgrim H, Diepstra A, Kamatani Y, Chabrier A, Gaborieau V, Boland A, Nieters A, Becker N, Foretova L, Benavente Y, Maynadié M, Staines A, Shield L, Lake A, Montgomery D, Taylor M, Smedby KE, Amini RM, Adami HO, Glimelius B, Feenstra B, Nolte IM, Visser L, van Imhoff GW, Lightfoot T, Cocco P, Kiemeney L, Vermeulen SH, Holcatova I, Vatten L, Macfarlane GJ, Thomson P, Conway DI, Benhamou S, Agudo A, Healy CM, Overvad K, Tjønneland A, Melin B, Canzian F, Khaw KT, Travis RC, Peeters PH, González CA, Quirós JR, Sánchez MJ, Huerta JM, Ardanaz E, Dorronsoro M, Clavel-Chapelon F, Bueno-de-Mesquita HB, Riboli E, Roman E, Boffetta P, de Sanjosé S, Zelenika D, Melbye M, van den Berg A, Lathrop M, Brennan P, McKay JD. Genome-wide association study of classical Hodgkin lymphoma and Epstein-Barr virus status-defined subgroups. J Natl Cancer Inst. 2012; 104:240-53. http://dx.doi.org/10.1093/jnci/djr516 PMid:22286212 PMCid:PMC3274508  
  59. Hohaus S, Massini G, D'Alo' F, Guidi F, Putzulu R, Scardocci A, Rabi A, Di Febo AL, Voso MT, Leone G. Association between glutathione S-transferase genotypes and Hodgkin's lymphoma risk and prognosis. Clin Cancer Res. 2003; 9:3435-40. PMid:12960134       
  60. Hohaus S, Di Ruscio A, Di Febo A, Massini G, D'Alo' F, Guidi F, Mansueto G, Voso MT, Leone G. Glutathione S-transferase P1 genotype and prognosis in Hodgkin's lymphoma. Clin Cancer Res. 2005; 11:2175-9. http://dx.doi.org/10.1158/1078-0432.CCR-04-1250 PMid:15788664       
  61. Ribrag V, Koscielny S, Casasnovas O, Cazeneuve C, Brice P, Morschhauser F, Gabarre J, Stamatoullas A, Lenoir G, Salles G; Groupe d'Etude des Lymphomes agressifs group, Laboratoire de Génétique et de recherche translationnelle, and Institut Gustave Roussy. Pharmacogenetic study in Hodgkin lymphomas reveals the impact of UGT1A1 polymorphisms on patient prognosis. Blood. 2009; 113:3307-13. http://dx.doi.org/10.1182/blood-2008-03-148874 PMid:18768784       
  62. Lourenço GJ, Lorand-Metze I, Delamain MT, Miranda EC, Kameo R, Metze K, Lima CS. Polymorphisms of glutathione S-transferase mu 1, theta 1, and pi 1 genes and prognosis in Hodgkin lymphoma. Leuk Lymphoma. 2010; 51:2215-21. http://dx.doi.org/10.3109/10428194.2010.527402 PMid:20977336       
  63. Morabito F, Hohaus S, Mammi C, Marcheselli L, Gentile M, Merli F, Montanini A, Stelitano C, La Sala A, Scalone R, Voso MT, Luminari S, Iannitto E, Gobbi P, Federico M. Role of glutathione-S-transferase (GST) polymorphisms in patients with advanced Hodgkin lymphoma: results from the HD2000 GISL trial. Leuk Lymphoma. 2012;53:406-10. http://dx.doi.org/10.3109/10428194.2011.623254 PMid:21916526       
  64. Yri OE, Ekstrøm PO, Hilden V, Gaudernack G, Liestøl K, Smeland EB, Holte H. Polymorphisms in genes encoding interleukin-10 and drug metabolizing enzymes GSTP1, GSTT1, GSTA1 and UGT1A1 influence risk and outcome in Hodgkin lymphoma. Leuk Lymphoma. 2012; 53:1934-44. http://dx.doi.org/10.3109/10428194.2012.682307 PMid:22475179       
  65. Schoof N, Franklin J, Fürst R, Zander T, von Bonin F, Peyrade F, Trümper L, Diehl V, Engert A, Kube D, Re D. Interleukin-10 gene polymorphisms are associated with freedom from treatment failure for patients with Hodgkin lymphoma. Oncologist. 2013;18:80-9. http://dx.doi.org/10.1634/theoncologist.2012-0291 PMid:23299779 PMCid:PMC3556261  
  66. Ghesquières H, Maurer MJ, Casasnovas O, Ansell SM, Larrabee BR, Lech-Maranda E, Novak AJ, Borrel AL, Slager SL, Brice P, Allmer C, Brion A, Ziesmer SC, Morschhauser F, Habermann TM, Gaillard I, Link BK, Stamatoullas A, Fermé C, Dogan A, Macon WR, Audouin J, Cerhan JR, Salles G. Cytokine gene polymorphisms and progression-free survival in classical Hodgkin lymphoma by EBV status: results from two independent cohorts. Cytokine. 2013; 64:523-31. http://dx.doi.org/10.1016/j.cyto.2013.08.002 PMid:24008079       
  67. Josting A, Franklin J, May M, Koch P, Beykirch MK, Heinz J, Rudolph C, Diehl V, Engert A. New prognostic score based on treatment outcome of patients with relapsed Hodgkin's lymphoma registered in the database of the German Hodgkin's lymphoma study group. J Clin Oncol. 2002; 20:221-30. http://dx.doi.org/10.1200/JCO.20.1.221 PMid:11773173       
  68. Hohaus S, Giachelia M, Di Febo A, Martini M, Massini G, Vannata B, D'Alo' F, Guidi F, Greco M, Pierconti F, Larocca LM, Voso MT, Leone G. Polymorphism in cytokine genes as prognostic markers in Hodgkin's lymphoma. Ann Oncol. 2007;18:1376-81. http://dx.doi.org/10.1093/annonc/mdm132 PMid:17496310       
  69. Moskowitz CH, Nimer SD, Zelenetz AD, Trippett T, Hedrick EE, Filippa DA, Louie D, Gonzales M, Walits J, Coady-Lyons N, Qin J, Frank R, Bertino JR, Goy A, Noy A, O'Brien JP, Straus D, Portlock CS, Yahalom J. A 2-step comprehensive high-dose chemoradiotherapy second-line program for relapsed and refractory Hodgkin disease: analysis by intent to treat and development of a prognostic model. Blood. 2001; 97:616-23. http://dx.doi.org/10.1182/blood.V97.3.616 PMid:11157476        

[TOP]